You are on page 1of 12

GASTROENTEROLOGY 2009;136:20032014

The Relationship Between Intestinal Microbiota and the Central Nervous


System in Normal Gastrointestinal Function and Disease

Stephen M. Collins Premysl Bercik


The Farncombe Family Digestive Health Research Institute, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada

Although many people are aware of the communica- consider the influence of the brain on the microbial content
tion that occurs between the gastrointestinal (GI) of the gut and, conversely, to examine the evidence showing
tract and the central nervous system, fewer know that the intestinal microbiota influences the brain and be-
about the ability of the central nervous system to havior. Investigation of the integration of the intestinal
influence the microbiota or of the microbiotas influ- microbiota into the GBA could improve the understanding
ence on the brain and behavior. Within the GI tract, of the pathophysiology of both functional1 and inflamma-
the microbiota have a mutually beneficial relation- tory2 bowel conditions.
ship with their host that maintains normal mucosal The GBA contributes to homeostasis of several sys-
immune function, epithelial barrier integrity, motil- tems, including GI function, appetite, and weight con-
ity, and nutrient absorption. Disruption of this rela- trol. Because GI motility and epithelial function are crit-
tionship alters GI function and disease susceptibility. ical determinants of the habitat for the microbiota,
Animal studies suggest that perturbations of behav- changes induced by the central nervous system or the GI
ior, such as stress, can change the composition of the tract alter the habitat and perturb the intestinal micro-
microbiota; these changes are associated with in- biota.3 The longstanding observation that oral antibiot-
creased vulnerability to inflammatory stimuli in the ics and laxatives ameliorate hepatic encephalopathy pro-
GI tract. The mechanisms that underlie these alter- vides a potent reminder that the intestinal microbiota is
ations are likely to involve stress-induced changes in capable of influencing behavior, albeit under pathologic
GI physiology that alter the habitat of enteric bacte- conditions.4 Taken together, these observations provide a
ria. Furthermore, experimental perturbation of the framework for considering the integration of the intesti-
microbiota can alter behavior, and the behavior of nal microbiota into the bidirectional GBA.
germ-free mice differs from that of colonized mice.
Gaining a better understanding of the relationship The Intestinal Microbiota
between behavior and the microbiota could provide
The gut contains a vast and complex microbial
insight into the pathogenesis of functional and in-
ecosystem, comprising mainly bacteria, of which most are
flammatory bowel disorders.
strict anaerobes; it also includes fungi and viruses,57 but
only bacteria are considered in this review. Commensal

T he gut brain axis (GBA) is a bidirectional neurohu-


moral communication system that integrates brain
and gastrointestinal (GI) functions. The GBA has been im-
bacteria instruct the immune and physiologic systems
throughout life and are responsible for the presence of
inflammatory and immune cells in the healthy gut: so-
plicated in the pathophysiology of functional GI disorders, called physiologic or controlled inflammation. The
and evidence is emerging for its role in the pathogenesis of term physiologic inflammation refers to the presence of
inflammatory disorders of the gut such as inflammatory
bowel disease (IBD). It would be a relatively straightforward Abbreviations used in this paper: ACTH, adrenocorticotrophic hor-
matter to integrate information about the intestinal micro- mone; GBA, gut brain axis; GI, gastrointestinal; IBD, inammatory
biota with that of the GBA by simply reviewing literature on bowel disease; IBS, irritable bowel syndrome; SPF, specic pathogen-
free.
interactions between flora and the GI tract. However, the 2009 by the AGA Institute
brain is the most influential organ within the axis, and 0016-5085/09/$36.00
communication is bidirectional. Thus, it is important to doi:10.1053/j.gastro.2009.01.075
2004 COLLINS AND BERCIK GASTROENTEROLOGY Vol. 136, No. 6

inflammatory cells in the mucosa and submucosa of the lymphoid structures are not developed. The significant
healthy GI tract and reflects the presence and immuno- number of inflammatory cells in the lamina propria of
logic accommodation (rather than immune tolerance) of the colonized intestine of healthy hosts and the preser-
the intestinal microbiota. The microbiota serves the host vation of normal epithelial structure and function are
by protecting against pathogens, participating in the reflections of the delicate and mutually beneficial rela-
intake nutrients from the diet, metabolizing certain tionship between the intestinal microbiota and the host.
drugs and carcinogens, and influencing the absorption Disruption of this balance, as a result of perturbation of
and distribution of fat.8,9 The influence of the intestinal the microbiota by infection or antibiotics, results in dys-
microbiota extends beyond the GI tract, contributing to, biosis. The effect of dysbiosis on the host is determined
for example, pain perception in the skin10 and fat depo- by the nature and magnitude of change in the bacteria
sition in the liver.11,12 Disruption of the symbiotic rela- composition of the GI tract, as well as by host suscepti-
tionship between the microbiota and the GI tract, re- bilities.
ferred to as dysbiosis,13 perturbs host functions and, in Another strategy for assessing the effect of the intesti-
some cases, causes the expression of overt and serious nal microbiota on host function is to perturb the com-
diseases such as IBD and Clostridium difficile colitis.14 16 mensal bacteria with the use of oral antibiotics.28 A
combination of neomycin and bacitracin altered the mi-
Influence of the Microbiota on the GI crobiota in mice, substantially reducing the Lactobacillus
Tract population.29 As shown in Figure 1, this resulted in a
A strategy that is commonly used to investigate small increment in myeloperoxidase (MPO) activity (a
interactions between the microbiota and the host is to measure of granulocytic inflammatory cell activity) with-
compare germ-free animals with those colonized with a out causing tissue damage. This increment in physiologic
single strain or multiple strains of bacteria.17 The micro- inflammation was accompanied by an increase in immu-
biota influences expression of a broad array of host genes. noreactive substance P, a sensory neurotransmitter, in
A comparison of germ-free mice and mice colonized with the intestinal wall. The functional consequence was an
Bacteroides thetaiotaomicron, a prominent member of the increase in the visceromotor or pseudoaffective response
adult mouse and human gut microflora, showed that the (abdominal wall contraction after colorectal balloon dis-
microbiota modulate the expression of genes that regu- tension), a widely used measure of visceral pain.30 Thus,
late nutrient absorption, mucosal barrier enhancement, perturbation of the microbiota produced a response pro-
xenobiotic metabolism, and angiogenesis.18 Colonization file reminiscent of changes seen in some patients with
with B. thetaiotaomicron also induced a 2- to 5-fold in- irritable bowel syndrome (IBS): subclinical inflammation
crease in mRNA encoding the synaptic vesicle-associated or immune activation and visceral hyperalgesia. Interest-
protein-33,18 which is involved in synaptic neurotrans- ingly, when the mice were gavaged with Lactobacillus pa-
mission.19 This finding indicates that commensal bacte- racasei, the antibiotic-induced changes in inflammation,
ria can influence the expression of genes whose products neurotransmitter content, and the visceromotor response
influence function in the nervous system. improved.29 Because the antibiotic-induced changes in
Comparisons of germ-free and colonized animals indi- the visceromotor response, immunoreactive substance P,
cate that, although crypt villous formation does not and myeloperoxidase activity could also be attenuated by
require the presence of bacteria, epithelial cell differenti- the administration of dexamethasone, it was concluded
ation, including Paneth cell development, depends on the that the changes in visceral perception were secondary to
microbiota in a way that serves both the host and resi- the increase in the inflammatory or immune cell presence
dent bacteria (for review, see Falk et al17). Similarly, the induced by the dysbiosis.29 These results show that com-
production and composition of mucin20 and the devel- mensal bacteria can influence primary afferent nerves in
opment of 5-hydoxy-tryptaminesecreting enteroendo- the gut and serve as an example of a functional relation-
crine cells is influenced by the microbiota.21 Germ-free ship between the sensory component of nervous system
rodents have an enlarged cecum, reflecting a gross dis- and the intestinal microbiota.
turbance in GI motility22,23; its prompt reversal to normal
size on bacterial colonization identifies the microbiota as Influence of GI Physiology on the
a determinant of GI motility.24 26 The abnormal motility Microbiota
of germ-free animals probably reflects a combination of Although the microbiota exert a broad influence
the lack of a mature enteroendocrine system,21 changes on host physiology, the converse is also true. Under
in neurotransmission,18 and immaturity of the mucosal normal conditions, the GI tract provides a stable habitat
immune system. The intestinal microbiota also has an for commensal bacteria that supports its structural and
important influence on the imprinting, maturation, and functional integrity (Figure 2A). Disturbance of normal
maintenance of the mucosal immune system (for reviews, GI physiology destabilizes the habitat, resulting in
see Falk et al17 and Macpherson et al27). Inflammatory changes in its microbial composition. An example of this
cells are sparse in the germ-free intestine, and secondary is the change in the bacterial composition of the GI tract
May 2009 GUT MICROBES AND THE BRAIN 2005

Figure 1. The effect of antibiotic-induced perturbation of the intestinal microbiota on myeloperoxidase activity (A), immune-reactive substance P in
the intestinal wall (B), and visceromotor response to balloon distension in mice (C) with or without treatment with Lactobacillus paracasei. AB,
antibiotic; ATB, antibiotic; AUC, area under the curve; Con, control; CRD, colorectal distension; LB, lactobacillus. Adapted with permission from
Verdu et al.29

after interruption of normal interdigestive motility in the ders would stabilize both host physiology and the bacte-
rat31 and in human beings.3 Changes in epithelial cell rial composition of the GI tract.
physiology, mucous secretion, and intestinal barrier func-
tion are also likely to affect the mucosal-associated mi- Ability of the Brain to Influence
crobial ecosystem.17 The sympathetic nervous system fa- Microbiota
cilitates the selective presentation of enteric bacteria to Several animal studies suggest that psychological
the mucosal immune system.32 Norepinephrine-contain- stress alters GI flora, but each of these studies has limi-
ing nerve fibers were identified in close proximity to the tations. Tannock and Savage36 reported changes in GI
epithelium that overlies lymphoid follicles in pig jeju- flora of mice stressed by deprivation of food, water, or
num. Application of norepinephrine increased the up- bedding. However, these environmental changes would
take of pathogenic bacteria into the follicles; this was be expected to have a direct effect on the microbiota that
prevented by the adrenergic antagonist phentolamine. is independent from a stress response. Bailey and Coe37
However, the underlying cellular mechanisms were not used maternal separation to demonstrate a reduction in
elucidated but probably involve the interaction of sym- lactobacilli for 7 days in infant rhesus monkeys; these
pathetic nerves with dendritic cells or specialized epithe- results should be interpreted cautiously because the stres-
lium to sample luminal bacteria and selectively take up sor limited maternal contact, and the analysis was limited
pathogenic bacteria for presentation to the mucosal im- to the identification of culturable bacteria. An interesting
mune system.32 finding was that the reduction in lactobacilli appeared to
Evidence also suggests that the release of biologic promote the emergence of enteric pathogens such as
amines, such as norepinephrine, can influence the com- Campylobacter jejuni. Additional studies showed the ability
position of the intestinal microbiota. This neurotrans- of probiotics to ameliorate stress-induced changes in GI
mitter has been shown to stimulate the growth of patho- function38 and to attenuate the observed reduction in
genic and nonpathogenic Escherichia coli in vitro and to lactobacilli in maternally deprived rat pups.39 A recent
influence their adherence to the mucosa.3335 Changes in study showed that stress during early life (maternal sep-
host physiology, initiated within the GI tract or by the aration) produced changes in the microbiota of the off-
central nervous system, produce changes in the bacterial spring, and this was associated with increased levels of
composition of the GI tract. Alternatively, changes in the corticosterone and inflammatory cytokines40; the inves-
microbiota, induced by infection or antibiotics, or other tigators speculated that this could reflect cytokine-in-
events such as stress (see below) perturb physiologic duced hyperresponsiveness of the hypothalamicpitu-
inflammation and GI physiology. A change in GI physi- itary pathway in response to maternal separation. In
ology provides an altered habitat that in turn supports a another study, maternal separation in mice was accom-
different microbiota. We propose that this cycle (Figure panied by an increase in intestinal permeability and a
2B) could be a basis for maintaining a state of GI dys- vulnerability of the GI tract to inflammatory stimuli.41
function after perturbation of the microbiota; it could Taken together, research conducted on offspring that
also explain the development and persistence of dysbiosis had been separated from their mothers show an in-
in conditions in which there is a primary disturbance of creased stress response, a systemic cytokine response,
GI physiology. So, the optimal therapy for these disor- increased intestinal permeability, and a shift in the bac-
2006 COLLINS AND BERCIK GASTROENTEROLOGY Vol. 136, No. 6

Figure 2. (A) The interrelationship of the intestinal microbiota and gastrointestinal physiology and inflammation in health (A) and in the presence of
intestinal dysbiosis (B). (A) The gastrointestinal tract under normal conditions provides a stable habitat for commensal bacteria that supports the
structural and functional integrity of the gut. The stable bacterial population in turn supports normal gut physiology. (B) Disturbance of (1) normal gut
physiology or (2) the microbiota destabilizes the habitat, resulting in change in physiologic inflammation which in turn alters physiology.

terial composition of the GI tract; these changes could mechanistic basis for hepatic encephalopathy is incom-
contribute to the increased susceptibility of the GI tract pletely understood,45 there is some evidence from a rat
to chemical and infectious inflammatory stimuli ob- model of hepatic failure that certain bacteria can produce
served in stress models. Because the stressor, separation a ligand for the benzodiazepine receptor that may con-
of mother and offspring, is applied at a time when the GI tribute to the encephalopathy.46 Observations in human
tract is becoming colonized and host immune and phys- beings offer provocative but nevertheless weaker evidence
iological systems are maturing, the extent to which these of communication between the microbiota and brain.
findings can be applied to adult animals is limited. Re- Patients with symptoms of depression have been shown
serpine-induced depression in adult mice was also accom- to have abnormal profiles of breath hydrogen excretion
panied by a vulnerability to GI tract inflammation,42 but after ingesting fructose and other sugars.47 Eliminating
underlying mechanisms might differ from those ob- fructose from the diet resulted in an improvement in
served in the maternal separation model, and the effect of depression.48 In addition, fructose malabsorption was
the adult-onset depression on the microbiota has not yet accompanied by a reduction in plasma tryptophan.49
been studied. Fructose malabsorption provides substrate for rapid bac-
There are several mechanisms by which stress can alter terial fermentation, resulting in changes in GI motility,
the bacterial composition of the GI tract, including changes the mucosal biofilm, and the profile of the microbiota.50
in epithelial cell function and mucus secretion as well as A recent study showed that rats given Bifidobacteria infantis
changes in GI motility.41,43,44 As described, release of nor- for 14 days had increased plasma tryptophan levels, sug-
epinephrine into the GI tract during stress might preferen- gesting that commensal bacteria have the ability to in-
tially stimulate the growth of specific strains of bacteria as fluence tryptophan metabolism.51 Thus, it is possible
well as their ability to adhere to the mucosa.3234 that the reported linkages between carbohydrate malab-
sorption and depressive-like behavior reflect bacterial in-
Ability of Microbiota to Influence the terference with tryptophan metabolism.51 Culture- and
Brain and Behavior molecular-based analyses have shown changes in the mi-
The most compelling evidence of a GI microbe crobiota of a small number of autistic patients, compared
brain interaction is the often dramatic improvement in with controls, with a greater prevalence of clostridial
patients with hepatic encephalopathy after the adminis- species in autistic patients.52,53 In an uncontrolled trial,
tration of oral antibiotics and laxatives.4 Although the vancomycin provided transient symptomatic relief to a
May 2009 GUT MICROBES AND THE BRAIN 2007

limited number of children with late-onset autism.54


However, the relationship between the microbiota and
autistic behavior remains speculative.
Studies have shown that the brain responds to the
introduction of noninvasive pathogenic bacteria into the
cecum; brain stem nuclei are rapidly activated,55and there
is expression of anxiety-like behavior in mice.56,57 This
response is thought to be mediated by signals from the
afferent vagus nerve to the nucleus of the solitary tract
and the lateral parabrachial nucleus.57 No evidence of
inflammation was observed within the short time frame
of these studies.57,58 The composition of commensal bac-
teria was not assessed in these studies, but persistent and
major perturbations of the microbiota were unlikely,
given the short duration of the experiments and the
absence of an overt inflammatory response to pathogens,
which is critical for a sustained disruption of the micro-
biota.59,60 Figure 3. The effect of mild restraint stress on plasma adrenocortico-
Studies in which mice were chronically infected with trophic hormone (ACTH) concentrations in germ-free and colonized
Helicobacter pylori also show evidence of behavioral young mice. Adapted with permission from Sudo et al.64
changes. This infection produces changes in gastric phys-
iology that gradually improve after successful eradication
of H. pylori.61 These changes were accompanied by an showed that enteropathogenic E. coli could increase the
alteration in feeding behavior that persisted after eradica- stress response after mono-colonization of germ-free mice.
tion of infection and resolution of the changes in gastric This effect of E. coli appears to involve attachment of the
physiology. The persistent alteration in feeding behavior bacteria to epithelial cells, because the effect was not ob-
was accompanied by changes in the hypothalamic appetite- served after mono-colonization with mutant E. coli that
regulating peptide pro-opiomelanocortin.62 The mecha- lacked the translocation intimin receptor, which is critical
nisms that mediate changes in the brain and in behavior for the successful attachment to the epithelium. Interest-
during and after H. pylori infection are unknown but could ingly, the reversibility of the exaggerated stress response was
involve persistent immune activation in response to the observed only in very young mice, indicating that there is a
infection.63 A direct effect of H. pylori is unlikely, given the critical period in which the plasticity of the neural regula-
persistence of the behavioral changes long after eradication. tion of the stress response is sensitive to input from the
The microbiota were not characterized in this model, but it microbiota. Another important finding of Sudo et al64 was
is interesting that changes in brain chemistry and behavior the reduction in brain-derived neurotrophic factor expres-
were reversed by gavage of Lactobacillus rhamnosus and Lacto- sion and protein levels in the cortex and hippocampus of
bacillus helveticus.62 germ-free mice compared with SPF mice. The brain-derived
Few studies have examined the brain and behavior in neurotrophic factor regulates several aspects of brain activ-
germ-free hosts. However, a study by Sudo et al64 provided ity, including mood and cognitive function. In a preliminary
insight into the role of commensal bacteria in the imprint- report, McVeyNeufeld et al65 identified defects in contex-
ing of the hypothalamicpituitary response to stress. Dur- tual learning in germ-free mice under stress and nonstress
ing mild restraint stress, the investigators observed an conditions. They also found that germ-free mice exhibited
increase in adrenocorticotrophic hormone (ACTH) and higher levels of anxiety when stressed (an exaggerated stress
corticosterone release in young germ-free mice, compared response). Taken together, these reports demonstrate that
with young, colonized specific pathogen-free (SPF) mice the intestinal microbiota influence the development of
(Figure 3). The response observed in germ-free mice was brain responses to stress and influence cognitive function in
specific to the stressor and did not occur during ether- young mice.
induced stress. The increases in stress-induced ACTH and Experimental perturbation of the intestinal microbiota
corticosterone release were completely reversed when germ- influences behavior in adult mice (Figure 4). In that
free mice were colonized with B. infantis but only partially study,66 mice were given antibiotics (neomycin and bac-
reversed when germ-free mice were colonized with flora itracin) for 7 days by gavage, along with the antifungal
from SPF mice. This observation is important because it agent primaricin to perturb the microbiota.28,29 Changes
suggests that, within the flora of SPF mice, there are bacte- in behavior, as assessed by the step-down test and the
ria that contribute to suppression of the ACTH response light box dark box test, were observed in the antibiotic-
and bacteria that increase this response; B. infantis clearly treated mice; these tests measure anxiety-like behavior or
belongs to the former category. The investigators also timidity.67 Antibiotic-treated Balb/c mice showed a re-
2008 COLLINS AND BERCIK GASTROENTEROLOGY Vol. 136, No. 6

and might signal the presence of commensal bacteria to


the brain by the vagus nerve.71 There is close integration
of innate and adaptive responses, and the integrity of the
adaptive immune response is important for normal cog-
nitive function. Specifically, Kipnis et al73 showed that, in
mice, a deficit in peripheral T cells can result in cognitive
and behavioral impairment, although the origin of these
cells, and whether they cross the blood brain barrier or
signal from the periphery to influence behavior, is not
known.
The vagus nerve has an important role in signaling
from the GI tract to the brain and can be stimulated by
Figure 4. The effect of antibiotic-induced perturbation of the micro- bacteria products such as endotoxins or inflammatory
biota in mice. Mice were treated with bacitracin, neomycin, and prima- cytokines such as interleukin-1 and tumor necrosis fac-
ricin28 by gavage for 7 days. Behavior was assessed in the step-down tor .74 The vagal response to stimulation by peripheral
test (left), and in the light box dark box test (middle and right). Re- inflammatory events is the suppression of proinflamma-
sponses of control mice are shown in the yellow bars and those of
antibiotic-treated mice are shown in the blue bars. Antibiotic-treated
tory cytokine release from intestinal macrophages medi-
mice showed a significantly lower latency to step-down, greater time ated by the -7 subunit of the nicotinic acetylcholine
spent in the white box, and an increased number of crossovers be- receptor on these cell.74,75 Interestingly, this response is
tween the light and dark boxes. ATB, antibiotics. attenuated by the induction of depression in the
mouse.42 The introduction of noninvasive pathogens is
rapidly signaled to the brain, reflected by increased activ-
duced latency to step-down and increased activity in the ity of vagus nuclei in the brain stem, and this is accom-
light box dark box test (Figure 4). In addition, the mice panied by anxiety-like behavior in mice.55,57,58 It is possi-
given antibiotics spent more time exploring the light box, ble that perturbation of the microbiota is signaled in a
compared with control mice. These findings indicate that vagus-dependent manner, resulting in altered behavior.
perturbation of the microbial content of the GI tract in For example, introduction of lactobacilli to the duode-
adult mice results in measurable changes in behavior. num of rats has been shown to increase gastric vagal
The mice did not show fear or anxiety, but rather greater activity within minutes.76
activity after perturbation of the microbiota; this is not How is the presence of commensal bacteria communi-
consistent with a malaise effect of the gavaged antibiot- cated to the brain and how does it induce behavior
ics. On the basis of these findings, we propose that changes? Some studies indicate that soluble factors are
perturbation of the microbiota can influence behavior. involved. Factor-S is a sleep-inducing substance that ac-
This is consistent with a recent study reporting behav- cumulates in the brain and body fluids of sleep-deprived
ioral changes in mice in which the microbiota had been animals. It is unique because of its bacterial origin and is
perturbed by dietary alterations.68 derived from the bacterial cell wall. Studies suggest that
The mechanisms by which the microbiota influence GI bacteria are an important source of Factor-S because
behavior are unknown but could include immune-medi- normal sleep patterns were disrupted after perturbation
ated, neural, or humeral mechanisms. These mechanisms of the microbiota with oral antibiotics.76,77 Commensal
are by no means mutually exclusive; it is likely that they bacteria also produce precursors of benzodiazepine re-
occur in series or in parallel. Immune mechanisms in- ceptor ligands that could contribute to encephalopathy
clude activation of the innate immune response in the GI in a rat model of liver failure.46 Cognitive function im-
tract. Toll-like receptors-2, -4, and -5 are down-regulated proved in patients with minimal hepatic encephalopathy
in germ-free mice and become up-regulated during colo- given Bifidobacterium longum with fructo-oligosaccharide
nization, implying interactions between these receptors for 9 weeks. Although the mechanism of action is poorly
and the microbiota.69 Dendritic cells of the GI tract have understood, B. longum might inhibit the activity of ure-
processes that breach the epithelial layer and interact ase-positive commensal bacteria, which would reduce
with commensal bacteria to induce the production of ammonia levels or the production of other substances of
immunoglobulin A by B lymphocytes and plasma cells.70 bacterial origin, including mercaptans and thioles.78
Secreted immunoglobulin A limits the penetration of the
epithelium by the microbiota. These mechanisms restrict
the inflammatory response to commensal bacteria to the Probiotics
level seen under normal conditions (physiologic inflam- Probiotics are microbes (bacteria or yeast) that
mation). Dendritic cells are in close proximity to nerves confer health benefits to the host when administered in
in the GI tract71; the sensory neuropeptide calcitonin- sufficient quantity. They have been shown to influence
gene-related peptide modulates dendritic cell function72 function in a variety of organs, including the nervous
May 2009 GUT MICROBES AND THE BRAIN 2009

Figure 5. A hypothetical model describing the role of the intestinal microbiota in the pathogenesis of irritable bowel syndrome. In this model, known
risk factors for IBS that include gastroenteritis, antibiotic use, and stress produce changes in commensal bacteria and an increment in physiologic
inflammation. This leads to changes in gut function as a basis for abdominal symptom generation. Perturbation of the microbiota may contribute to
the behavioral profile seen in this condition.

system. Several studies report the effects of probiotics on acid in the probiotic-treated rats, compared with con-
the GBA and in models of altered behavior. trols. However, there was an unexplained reduction in the
In rats subjected to water-avoidance stress, intestinal concentration of 5-hydroxy-indole-acetic acid in the fron-
barrier function was reduced, and bacterial adherence to tal cortex and a decrease in dihydroxyphenylacetic acid, a
the epithelium was increased. In addition, commensal metabolite of dopamine, in the amygdaloid cortex in the
bacteria translocated across a leaky epithelial barrier, rats given the probiotics, and no improvement in the
ultimately reaching the mesenteric lymph nodes.79 Expo- forced swim test was observed. Although treatment failed
sure of rats to the probiotics L. helveticus and L. rhamnosus to influence behavior, the results of this study are impor-
prevented the stress-induced increase in adherence as well tant because they show the antidepressant potential of
as the translocation of commensal bacteria. Although it the bacterium B. infantis, primarily by virtue of its ability
was observed that the probiotics improved epithelial to increase the serotonergic precursor, tryptophan.51
function, it is not clear exactly how the probiotics func-
tioned in this model of the stress response.
Rat pups that experience maternal deprivation have a MicrobiotaGBA and Disease
significant increase in the visceromotor response to co- Evidence is increasing for a role of the GBA in the
lonic distension and increased GI paracellular permeabil- pathogenesis of IBD. Imbalance between the sympathetic
ity, compared with controls. The probiotic L. paracasei and parasympathetic outflow from the central nervous
significantly improved stress-induced visceral pain and system has been reported in patients with IBD81 83 and
restored normal GI permeability in the stressed rats. The may be associated with behavioral change. For example,
investigators concluded that this was mediated, at least depression has been correlated with Crohns disease,
in part, by a soluble factor that was derived from the stress, and ulcerative colitis in separate groups of pa-
probiotic. However, the mechanisms of action were not tients.84 A controlled study found an increased preva-
identified.80 lence of depression in patients with IBD.85 It is difficult
Another study investigated the antidepressant poten- to ascertain from human studies whether behavioral
tial of a probiotics B. infantis given for 14 days to rats that changes are primary or occur as a result of the morbidity
were chronically subjected to the forced swim test as a of these conditions. Animal studies show that stress ex-
stressor.51 This probiotic therapy resulted in a reduction acerbates experimental colitis and that depression in-
in levels of interferon , tumor necrosis factor , and creases susceptibility to inflammatory stimuli by impair-
interleukin-6 after mitogen stimulation of peripheral ing vagal parasympathetic outflow to the gut.41,42,86
blood monocytes. In addition, there was a marked and Given the experimental evidence that perturbation of the
significant increase in plasma tryptophan and kynurenic microbiota alters behavior and that dysbiosis occurs in
2010 COLLINS AND BERCIK GASTROENTEROLOGY Vol. 136, No. 6

Figure 6. The integration of the intestinal microbiota into the brain gut axis (GBA). Shown are the putative mechanisms whereby the brain may
influence the composition of the intestinal microbiota, and whereby the microbiota may influence the brain. The communication between these
systems is bidirectional. These are the components of the proposed bidirectional microbiota gut brain axis. CNS, central nervous system; EE,
entero-endocrine.

IBD, we speculate that dysbiosis could also contribute to IBS,104 but, unfortunately, the investigators did not cor-
the behavioral changes reported in some patients with relate these findings with symptom fluctuation. Never-
IBD. theless, taken together, these findings support the exis-
IBS is considered to be a disorder of the GBA, and tence of intestinal dysbiosis in IBS.
evidence is emerging of dysbiosis in patients with IBS. In animal studies, dysbiosis has been shown to induce
Factors that are known to predispose individuals to IBS low-grade inflammation that is not accompanied by tis-
include enteric infection, antibiotic use, and stress and sue damage,29 and it is plausible that dysbiosis is the
are also known to alter the bacterial composition of the cause of the low-grade inflammation found in mucosal
GI tract. Acute bacterial gastroenteritis is the strongest biopsies in subsets of patients with IBS. The longstand-
risk factor known for the development of IBS.8790 Anti- ing notion that IBS represents a low-grade inflammatory
biotic usage in children or in the context of postinfective disorder105 is now supported by several lines of evidence,
IBS is also a risk factor for the development of IBS.91,92 including genetic studies,106,107 semiquantitative histo-
Patients with IBS have an increased response to stress,93 logic studies,108 as well as studies reporting increased
and stress is also a predictor of IBS94 as well as a deter- mediator production from inflammatory cells in the GI
minant of symptom severity.95 wall109 and systemic circulation.110 We speculate that
Indirect evidence of dysbiosis in patients with IBS was dysbiosis is a determinant of immune activation and
based on the analysis of fermentation profiles on stool or low-grade inflammation in this subset of patients with
breath samples. Some patients with IBS have demonstra- IBS.
ble qualitative or quantitative changes in fermentation The potential for the microbiota to produce small
profiles compared with healthy controls,96 99 but the increments in physiologic inflammation and thereby per-
interpretation of these findings remains controver- turb GI and brain function prompts consideration of a
sial.100,101 With the use of 16s rRNA analysis of stool unifying hypothesis for IBS. Historically, IBS has been
samples, a recent study found significant changes, par- viewed as a psychosomatic disorder, with emphasis on
ticularly in Lactobacillus species, in patients with IBS pa- psychiatric comorbidity and symptom reporting by pa-
tients compared with controls.102 A reduction in lactoba- tients with IBS, a central model of the pathogenesis of
cilli was also observed in patients with diarrhea- IBS. During the past decade, another school of thought
predominant IBS, whereas patients with constipation- has emerged, implicating gastroenteritis and low-grade
predominant IBS had increases in Veillonella species.103 inflammation as mechanisms that underlie GI dysfunc-
Another study showed a greater temporal instability of tion and the symptoms of IBS, a peripheral model of IBS.
the microbiota during a 6-month period in patients with However, psychiatric comorbidity also occurs in patients
May 2009 GUT MICROBES AND THE BRAIN 2011

with postinfective IBS, and low-grade inflammation is 11. Dumas ME, Barton RH, Toye A, et al. Metabolic profiling reveals
found in patients with IBS with or without a history of a contribution of gut microbiota to fatty liver phenotype in insu-
lin-resistant mice. Proc Natl Acad Sci U S A 2006;103:12511
gastroenteritis. These observations prompt consideration
12516.
for a role of microbiotaGBA interactions in the patho- 12. Backhed F, Ding H, Wang T, et al. The gut microbiota as an
physiology of IBS, particularly in patients with demon- environmental factor that regulates fat storage. Proc Natl Acad
strable intestinal dysbiosis. Sci U S A 2004;101:15718 15723.
On the basis of the experimental data reviewed in 13. Hawrelak JA, Myers SP. The causes of intestinal dysbiosis: a
this article, we propose a model to incorporate intes- review. Altern Med Rev 2004;9:180 197.
14. Lepage P, Colombet J, Marteau P, Sime-Ngando T, Dore J,
tinal dysbiosis into a conceptual framework of IBS,
Leclerc M. Dysbiosis in inflammatory bowel disease: a role for
illustrated in Figure 5. In this model, recognized IBS bacteriophages? Gut 2008;57:424 425.
risk factors, such as acute gastroenteritis, antibiotic 15. Jacobs NF Jr. Antibiotic-induced diarrhea and pseudomembra-
therapy, or stress, produce intestinal dysbiosis and an nous colitis. Postgrad Med 1994;95:111120.
incremental increase in physiologic inflammation in 16. McFarland LV. Epidemiology, risk factors and treatments for
the colon that is subclinical but sufficient to alter antibiotic-associated diarrhea. Dig Dis 1998;16:292307.
17. Falk PG, Hooper LV, Midtvedt T, Gordon JI. Creating and main-
neuromuscular function (and thus produce GI symp- taining the gastrointestinal ecosystem: what we know and need
toms). In addition, intestinal dysbiosis may contribute to know from gnotobiology. Microbiol Mol Biol Rev 1998;62:
to the behavioral profile of patients with IBS. This 11571170.
construct requires testing in clinical studies. 18. Hooper LV, Wong MH, Thelin A, Hansson L, Falk PG, Gordon JI.
In conclusion, experimental data and clinical observa- Molecular analysis of commensal host-microbial relationships
in the intestine. Science 2001;291:881 884.
tions support the integration of the intestinal microbiota
19. Skehel PA, Armitage BA, Bartsch D, et al. Proteins functioning in
into the GBA (Figure 6). These include the bidirectional synaptic transmission at the sensory to motor synapse of Aply-
interactions between the microbiota and GI physiology sia. Neuropharmacology 1995;34:1379 1385.
and the associations between the microbiota and behav- 20. Wold JK, Khan R, Midtvedt T. Intestinal glycoproteins of germ-
ior. Future research should focus on the contributions of free rats. Chemical composition of intestinal and fecal mucus
immunologic, neural, and biochemical or metabolic from germfree rats fed a chemically defined diet. Acta Pathol
Microbiol Scand [B] Microbiol Immunol 1971;79:525530.
pathways to the microbiotaGBA relationship. A better
21. Uribe A, Alam M, Johansson O, Midtvedt T, Theodorsson E.
understanding of these relationships will improve our Microflora modulates endocrine cells in the gastrointestinal
understanding of functional and inflammatory condi- mucosa of the rat. Gastroenterology 1994;107:1259 1269.
tions of the GI tract and of hepatic encephalopathy. This 22. Abrams GD, Bishop JE. Effect of normal microbial flora on
knowledge may also prompt further exploration of the gastrointestinal motility. Proc Soc Exp Biol (N Y) 1967;126:
role of the intestinal microbiota in behavioral illnesses 301304.
23. Gustafsson BE, Midtvedt T, Strandberg K. Effects of microbial
such as depression.
contamination on the cecum enlargement of germfree rats.
Scand J Gastroenterol 1970;5:309 314.
References 24. Husebye E, Hellstrom PM, Sundler F, Chen J, Midtvedt T. Influ-
ence of microbial species on small intestinal myoelectric activity
1. Chang L, Sundaresh S, Elliott J, et al. Dysregulation of the
and transit in germ-free rats. Am J Physiol Gastrointest Liver
hypothalamic-pituitary-adrenal (HPA) axis in irritable bowel syn-
Physiol 2001;280:G368 G380.
drome. Neurogastroenterol Motil 2009 l21:149 159.
25. Husebye E, Hellstrom PM, Midtvedt T. Intestinal microflora stim-
2. Stasi C, Orlandelli E. Role of the brain-gut axis in the pathophys-
ulates myoelectric activity of rat small intestine by promoting
iology of Crohns disease. Dig Dis 2008;26:156 166.
3. Vantrappen G, Jannssens J, Hellemans J, Ghoos Y. The interdi- cyclic initiation and aboral propagation of migrating myoelectric
gestive motor complex of normal subjects and patients with complex. Dig Dis Sci 1994;39:946 956.
bacterial overgrowth of the small intestine. J Clin Invest 1977; 26. Caenepeel P, Janssens J, Vantrappen G, Eyssen H, Coremans
59:1158 1168. G. Interdigestive myoelectric complex in germ-free rats. Dig Dis
4. Morgan MY. The treatment of chronic hepatic encephalopathy. Sci 1989;34:1180 1184.
Hepatogastroenterology 1991;38:377387. 27. Macpherson AJ, Geuking MB, McCoy KD. Immune responses
5. Eckburg PB, Bik EM, Bernstein CN, et al. Diversity of the human that adapt the intestinal mucosa to commensal intestinal bac-
intestinal microbial flora. Science 2005;308:16351638. teria. Immunology 2005;115:153162.
6. OHara AM, Shanahan F. Gut microbiota: mining for therapeutic 28. van der WD, Sturm CA. Antibiotic decontamination of the diges-
potential. Clin Gastroenterol Hepatol 2007;5:274 284. tive tract of mice. Technical procedures. Lab Anim Care 1968;
7. Marchesi J, Shanahan F. The normal intestinal microbiota. Curr 18:110.
Opin Infect Dis 2007;20:508 513. 29. Verdu EF, Bercik P, Verma-Gandhu M, et al. Specific probiotic
8. Hooper LV, Gordon JI. Commensal host-bacterial relationships therapy attenuates antibiotic induced visceral hypersensitivity in
in the gut. Science 2001;292:11151118. mice. Gut 2006;55:182190.
9. Backhed F, Ley RE, Sonnenburg JL, Peterson DA, Gordon JI. 30. Burton MB, Gebhart GF. Effects of intracolonic acetic acid on
Host-bacterial mutualism in the human intestine. Science 2005; responses to colorectal distension in the rat. Brain Res 1995;
307:19151920. 672:7782.
10. Amaral FA, Sachs D, Costa VV, et al. Commensal microbiota is 31. Scott LD, Cahall DL. Influence of the interdigestive myoelectric
fundamental for the development of inflammatory pain. Proc complex on enteric flora in the rat. Gastroenterology 1982;82:
Natl Acad Sci U S A 2008;105:21932197. 737745.
2012 COLLINS AND BERCIK GASTROENTEROLOGY Vol. 136, No. 6

32. Green BT, Lyte M, Kulkarni-Narla A, Brown DR. Neuromodulation antidepressant properties in the rat. J Psychiatr Res 2008;43:
of enteropathogen internalization in Peyers patches from por- 164 174.
cine jejunum. J Neuroimmunol 2003;141:74 82. 52. Finegold SM, Molitoris D, Song Y, et al. Gastrointestinal micro-
33. Chen C, Brown DR, Xie Y, Green BT, Lyte M. Catecholamines flora studies in late-onset autism. Clin Infect Dis 2002;
modulate Escherichia coli O157:H7 adherence to murine cecal 35(Suppl):S6 S16.
mucosa. Shock 2003;20:183188. 53. Song Y, Liu C, Finegold SM. Real-time PCR quantitation of
34. Freestone PP, Williams PH, Haigh RD, Maggs AF, Neal CP, Lyte clostridia in feces of autistic children. Appl Environ Microbiol
M. Growth stimulation of intestinal commensal Escherichia coli 2004;70(11):6459 6465.
by catecholamines: a possible contributory factor in trauma- 54. Sandler RH, Finegold SM, Bolte ER, et al. Short-term benefit
induced sepsis. Shock 2002;18:465 470. from oral vancomycin treatment of regressive-onset autism.
35. Lyte M. The biogenic amine tyramine modulates the adherence J Child Neurol 2000;15(7):429 435.
of Escherichia coli O157:H7 to intestinal mucosa. J Food Prot 55. Wang X, Wang BR, Zhang XJ, Xu Z, Ding YQ, Ju G. Evidences for
2004;67:878 883. vagus nerve in maintenance of immune balance and transmis-
36. Tannock GW, Savage DC. Influences of dietary and environmen- sion of immune information from gut to brain in STM-infected
tal stress on microbial populations in the murine gastrointesti- rats. World J Gastroenterol 2002;8:540 545.
nal tract. Infect Immun 1974;9:591598. 56. Goehler LE, Lyte M, Gaykema RP. Infection-induced viscerosensory
37. Bailey MT, Coe CL. Maternal separation disrupts the integrity of signals from the gut enhance anxiety: implications for psychoneu-
the intestinal microflora in infant rhesus monkeys. Dev Psycho- roimmunology. Brain Behav Immun 2007;21:721726.
biol 1999;35:146 155. 57. Gaykema RP, Goehler LE, Lyte M. Brain response to cecal
38. Eutamene H, Bueno L. Role of probiotics in correcting abnor- infection with Campylobacter jejuni: analysis with Fos immuno-
malities of colonic flora induced by stress. Gut 2007;56:1495 histochemistry. Brain Behav Immun 2004;18:238 245.
1497. 58. Lyte M, Li W, Opitz N, Gaykema RP, Goehler LE. Induction of
39. Gareau MG, Jury J, MacQueen G, Sherman PM, Perdue MH. anxiety-like behavior in mice during the initial stages of infection
Probiotic treatment of rat pups normalises corticosterone re- with the agent of murine colonic hyperplasia Citrobacter roden-
lease and ameliorates colonic dysfunction induced by maternal tium. Physiol Behav 2006;89:350 357.
separation. Gut 2007;56:15221528. 59. Stecher B, Robbiani R, Walker AW, et al. Salmonella enterica
40. OMahony SM, Marchesi JR, Scully P, et al. Early life stress serovar Typhimurium exploits inflammation to compete with the
alters behavior, immunity, and microbiota in rats: implications intestinal microbiota. PLoS Biol 2007;5:21772189.
for irritable bowel syndrome and psychiatric illnesses. Biol Psy- 60. Lupp C, Robertson ML, Wickham ME, et al. Host-mediated
chiatry 2009;65:263267. inflammation disrupts the intestinal microbiota and promotes
41. Varghese AK, Verdu EF, Bercik P, et al. Antidepressants atten- the overgrowth of Enterobacteriaceae. Cell Host Microbe 2007;
uate increased susceptibility to colitis in a murine model of 2:204.
depression. Gastroenterology 2006;130:17431753. 61. Bercik P, De Giorgio R, Blennerhassett P, Verdu EF, Barbara G,
42. Ghia JE, Blennerhassett P, Collins SM. Impaired parasympa- Collins SM. Immune-mediated neural dysfunction in a murine
thetic function increases susceptibility to inflammatory bowel model of chronic Helicobacter pylori infection. Gastroenterology
disease in a mouse model of depression. J Clin Invest 2008; 2002;123:12051215.
118:2209 2218. 62. Bercik P, Verdu EF, Foster JA, et al. Role of gut-brain axis in
43. Groot J, Bijlsma P, Van Kalkeren A, Kiliaan A, Saunders P, persistent abnormal feeding behavior in mice following eradica-
Perdue M. Stress-induced decrease of the intestinal barrier tion of Helicobacter pylori infection. Am J Physiol Regul Integr
function. The role of muscarinic receptor activation. Ann N Y Comp Physiol 2009;296:R587R594.
Acad Sci 2000;915:237246. 63. Verdu EF, Bercik P, Huang XX, et al. The role of luminal factors
44. Rubio CA, Huang CB. Quantification of the sulphomucin-produc- in the recovery of gastric function and behavioral changes after
ing cell population of the colonic mucosa during protracted chronic Helicobacter pylori infection. Am J Physiol Gastrointest
stress in rats. In Vivo 1992;6:81 84. Liver Physiol 2008;295:G664 G670.
45. Williams R. Review article: bacterial flora and pathogenesis in 64. Sudo N, Chida Y, Aiba Y, et al. Postnatal microbial colonization
hepatic encephalopathy. Aliment Pharmacol Ther 2007;25(Suppl programs the hypothalamic-pituitary-adrenal system for stress
1):1722. response in mice. J Physiol 2004;558:263275.
46. Yurdaydin C, Walsh TJ, Engler HD, et al. Gut bacteria provide 65. McVeyNeufeld KA, Bienenstock J, JA Foster. The impact of
precursors of benzodiazepine receptor ligands in a rat model of intestinal microbiota on anxiety like behavior [abstract]. Neuro-
hepatic encephalopathy. Brain Res 1995;679:42 48. gastroenterol Motil 2008;20(Suppl2):125.
47. Ledochowski M, Widner B, Sperner-Unterweger B, Propst T, 66. Denou E, Bercik P, Collins SM. Perturbation of the intestinal
Vogel W, Fuchs D. Carbohydrate malabsorption syndromes and microbiota alters behavior in mice. Gastroenterology DDW (in
early signs of mental depression in females. Dig Dis Sci 2000; press).
45:12551259. 67. Crawley JN. Behavioral phenotyping of rodents. Comp Med
48. Ledochowski M, Widner B, Bair H, Probst T, Fuchs D. Fructose- 2003;53:140 146.
and sorbitol-reduced diet improves mood and gastrointestinal 68. Li W, Dowd SE, Scurlock B, Acosta-Martinez V, Lyte M. Memory
disturbances in fructose malabsorbers. Scand J Gastroenterol and learning behavior in mice is temporally associated with
2000;35:1048 1052. diet-induced alterations in gut bacteria. Physiol Behav 2009;96:
49. Ledochowski M, Widner B, Murr C, Sperner-Unterweger B, Fuchs D. 557567.
Fructose malabsorption is associated with decreased plasma tryp- 69. Lundin A, Bok CM, Aronsson L, et al. Gut flora, Toll-like receptors
tophan. Scand J Gastroenterol 2001;36(4):367371. and nuclear receptors: a tripartite communication that tunes in-
50. Gibson PR, Newnham E, Barrett JS, Shepherd SJ, Muir JG. nate immunity in large intestine. Cell Microbiol 2008;10:1093
Review article: fructose malabsorption and the bigger picture. 1103.
Aliment Pharmacol Ther 2007;25(4):349 363. 70. Macpherson AJ, Slack E. The functional interactions of commen-
51. Desbonnet L, Garrett L, Clarke G, Bienenstock J, Dinan TG. The sal bacteria with intestinal secretory IgA. Curr Opin Gastroen-
probiotic Bifidobacteria infantis: an assessment of potential terol 2007;23:673 678.
May 2009 GUT MICROBES AND THE BRAIN 2013

71. Goehler LE, Gaykema RP, Nguyen KT, et al. Interleukin-1beta in 89. Spiller RC. Estimating the importance of infection in IBS. Am J
immune cells of the abdominal vagus nerve: a link between the Gastroenterol 2003;98:238 241.
immune and nervous systems? J Neurosci 1999;19:2799 2806. 90. Rodriguez LA, Ruigomez A. Increased risk of irritable bowel
72. Hosoi J, Murphy GF, Egan CL, et al. Regulation of Langerhans syndrome after bacterial gastroenteritis: cohort study. BMJ
cell function by nerves containing calcitonin gene-related pep- 1999;318:565566.
tide. Nature 1993;363:159 163. 91. Ruigomez A, Garcia Rodriguez LA, Panes J. Risk of irritable
73. Kipnis J, Cohen H, Cardon M, Ziv Y, Schwartz M. T cell deficiency bowel syndrome after an episode of bacterial gastroenteritis in
leads to cognitive dysfunction: implications for therapeutic vac- general practice: influence of comorbidities. Clin Gastroenterol
cination for schizophrenia and other psychiatric conditions. Proc Hepatol 2007;5:465 469.
Natl Acad Sci U S A 2004;101:8180 8185. 92. Mendall MA, Kumar D. Antibiotic use, childhood affluence and
74. Borovikova LV, Ivanova S, Zhang M, et al. Vagus nerve stimula- irritable bowel syndrome (IBS). Eur J Gastroenterol Hepatol
tion attenuates the systemic inflammatory response to endo- 1998;10:59 62.
toxin. Nature 2000;405:458 462. 93. Mayer EA, Naliboff BD, Chang L, Coutinho SV. V. Stress and
75. Ghia JE, Blennerhassett P, Kumar-Ondiveeran H, Verdu EF, Col- irritable bowel syndrome. Am J Physiol Gastrointest Liver Physiol
lins SM. The vagus nerve: a tonic inhibitory influence associated 2001;280:G519 G524.
with inflammatory bowel disease in a murine model. Gastroen- 94. Fujii Y, Nomura S. A prospective study of the psychobehavioral
terology 2006;131:11221130. factors responsible for a change from non-patient irritable bowel
76. Tanida M, Yamano T, Maeda K, Okumura N, Fukushima Y, Nagai K. syndrome to IBS patient status. Biopsychosoc Med 2008;2:16.
Effects of intraduodenal injection of Lactobacillus johnsonii La1 on 95. Blanchard EB, Lackner JM, Jaccard J, et al. The role of stress in
renal sympathetic nerve activity and blood pressure in urethane- symptom exacerbation among IBS patients. J Psychosom Res
anesthetized rats. Neurosci Lett 2005;389(2):109 114. 2008;64:119 128.
77. Brown R, Price RJ, King MG, Husband AJ. Are antibiotic effects 96. King TS, Elia M, Hunter JO. Abnormal colonic fermentation in
on sleep behavior in the rat due to modulation of gut bacteria? irritable bowel syndrome. Lancet 1998;352:11871189.
Physiol Behav 1990;48:561565. 97. Lin HC. Small intestinal bacterial overgrowth: a framework for
78. Malaguarnera M, Greco F, Barone G, Gargante MP, Malaguarn- understanding irritable bowel syndrome. JAMA 2004;292:852
era M, Toscano MA. Bifidobacterium longum with fructo-oligo- 858.
saccharide (FOS) treatment in minimal hepatic encephalopathy:
98. Pimentel M, Lezcano S. Irritable bowel syndrome: bacterial over-
a randomized, double-blind, placebo-controlled study. Dig Dis
growthwhats known and what to do. Curr Treat Options Gas-
Sci 2007;52:3259 3265.
troenterol 2007;10:328 337.
79. Zareie M, Johnson-Henry K, Jury J, et al. Probiotics prevent
99. Treem WR, Ahsan N, Kastoff G, Hyams JS. Fecal short-chain
bacterial translocation and improve intestinal barrier function in
fatty acids in patients with diarrhea-predominant irritable bowel
rats following chronic psychological stress. Gut 2006;55:1553
syndrome: in vitro studies of carbohydrate fermentation. J Pe-
1560.
diatr Gastroenterol Nutr 1996;23:280 286.
80. Eutamene H, Lamine F, Chabo C, et al. Synergy between Lac-
100. Dear KL, Elia M, Hunter JO. Do interventions which reduce
tobacillus paracasei and its bacterial products to counteract
colonic bacterial fermentation improve symptoms of irritable
stress-induced gut permeability and sensitivity increase in rats.
bowel syndrome? Dig Dis Sci 2005;50:758 766.
J Nutr 2007;137:19011907.
101. Vanner S. The small intestinal bacterial overgrowth. Irritable
81. Lindgren S, Stewenius J, Sjlund K, Lilja B, Sundkvist G. Auto-
bowel syndrome hypothesis: implications for treatment. Gut
nomic vagal nerve dysfunction in patients with ulcerative colitis.
2008;57:13151321.
Scand J Gastroenterol 1993;28:638 642.
102. Kassinen A, Krogius-Kurikka L, Makivuokko H, et al. The fecal
82. Lindgren S, Lilja B, Rosn I, Sundkvist G. Disturbed autonomic
nerve function in patients with Crohns disease. Scand J Gas- microbiota of irritable bowel syndrome patients differs signifi-
troenterol 1991;26:361366. cantly from that of healthy subjects. Gastroenterology 2007;
83. Ganguli SC, Kamath MV, Redmond K, et al. A comparison of 133:24 33.
autonomic function in patients with inflammatory bowel disease 103. Malinen E, Rinttila T, Kajander K, et al. Analysis of the fecal
and in healthy controls. Neurogastroenterol Motil 2007;19:961 microbiota of irritable bowel syndrome patients and healthy
967. controls with real-time PCR. Am J Gastroenterol 2005;100:
84. Maunder RG, Levenstein S. The role of stress in the develop- 373382.
ment and clinical course of inflammatory bowel disease: epide- 104. Maukonen J, Satokari R, Matto J, Soderlund H, Mattila-Sandholm
miological evidence. Curr Mol Med 2008;8:247252. T, Saarela M. Prevalence and temporal stability of selected clos-
85. Walker JR, Ediger JP, Graff LA, et al. The Manitoba IBD cohort tridial groups in irritable bowel syndrome in relation to predominant
study: a population-based study of the prevalence of lifetime faecal bacteria. J Med Microbiol 2006;55:625 633.
and 12-month anxiety and mood disorders. Am J Gastroenterol 105. Collins SM. Is the irritable gut an inflamed gut? Scand J Gas-
2008;103:1989 1997. troent Suppl 1992;192:102105.
86. Qiu BS, Vallance BA, Blennerhassett PA, Collins SM. The role of 106. Gonsalkorale WM, Perrey C, Pravica V, Whorwell PJ, Hutchin-
CD4 lymphocytes in the susceptibility of mice to stress-in- son IV. Interleukin 10 genotypes in irritable bowel syndrome:
duced reactivation of experimental colitis. Nat Med 1999;5: evidence for an inflammatory component? Gut 2003;52:
1178 1182. 9193.
87. Marshall JK, Thabane M, Garg AX, Clark WF, Salvadori M, Collins 107. van der Veek PP, van den BM, de Kroon YE, Verspaget HW,
SM. Incidence and epidemiology of irritable bowel syndrome Masclee AA. Role of tumor necrosis factor-alpha and interleu-
after a large waterborne outbreak of bacterial dysentery. Gas- kin-10 gene polymorphisms in irritable bowel syndrome. Am J
troenterology 2006;131:445 450. Gastroenterol 2005;100:2510 2516.
88. Parry SD, Stansfield R, Jelley D, et al. Does bacterial gastroen- 108. Barbara G, Stanghellini V, De Giorgio R, et al. Activated mast
teritis predispose people to functional gastrointestinal disor- cells in proximity to colonic nerves correlate with abdominal
ders? A prospective, community-based, case-control study. Am J pain in irritable bowel syndrome. Gastroenterology 2004;
Gastroenterol 2003;98:1970 1975. 126:693702.
2014 COLLINS AND BERCIK GASTROENTEROLOGY Vol. 136, No. 6

109. Barbara G, Wang B, Stanghellini V, et al. Mast cell-dependent Acknowledgments


excitation of visceral-nociceptive sensory neurons in irritable The authors thank Dr Elena F. Verdu, Emmanuel Denou, PhD, and
bowel syndrome. Gastroenterology 2007;132:26 37. Jean-Eric Ghia, PhD, for their contributions to the work shown in this
110. OMahony L, McCarthy J, Kelly P, et al. Lactobacillus and bi- manuscript.
fidobacterium in irritable bowel syndrome: symptom responses
and relationship to cytokine profiles. Gastroenterology 2005; Conicts of interest
128:541551. The authors disclose the following: Dr S.M. Collins received a
grant in aid from the Nestle Research Institute, Switzerland, and
from Institut Rossel, Montreal, Canada. Dr P. Bercik received a
Received November 7, 2008. Accepted January 25, 2009. grant in aid from the Nestle Research Institute, Switzerland.

Reprint requests Funding


Address requests for reprints to: Stephen M. Collins, MB.BS, The work was supported primarily by the Canadian Institutes of
FRCPC, Faculty of Health Sciences, Room 2E17, McMaster Health Research (CIHR) and by unrestricted grants from the Nestle
University, 1200 Main Street West, Hamilton, ON, Canada L8N 3Z5. Research Institute, Switzerland, and Institut Rosell, Montreal,
e-mail: scollins@mcmaster.ca; fax: (905) 524-1346. Canada.

You might also like